1
|
Fan T, Xie J, Huang G, Li L, Zeng X, Tao Q. PHF8/KDM7B: A Versatile Histone Demethylase and Epigenetic Modifier in Nervous System Disease and Cancers. EPIGENOMES 2024; 8:36. [PMID: 39311138 PMCID: PMC11417953 DOI: 10.3390/epigenomes8030036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/23/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024] Open
Abstract
Many human diseases, such as malignant tumors and neurological diseases, have a complex pathophysiological etiology, often accompanied by aberrant epigenetic changes including various histone modifications. Plant homologous domain finger protein 8 (PHF8), also known as lysine-specific demethylase 7B (KDM7B), is a critical histone lysine demethylase (KDM) playing an important role in epigenetic modification. Characterized by the zinc finger plant homology domain (PHD) and the Jumonji C (JmjC) domain, PHF8 preferentially binds to H3K4me3 and erases repressive methyl marks, including H3K9me1/2, H3K27me1, and H4K20me1. PHF8 is indispensable for developmental processes and the loss of PHF8 enzyme activity is linked to neurodevelopmental disorders. Moreover, increasing evidence shows that PHF8 is highly expressed in multiple tumors as an oncogenic factor. These findings indicate that studying the role of PHF8 will facilitate the development of novel therapeutic agents by the manipulation of PHF8 demethylation activity. Herein, we summarize the current knowledge of PHF8 about its structure and demethylation activity and its involvement in development and human diseases, with an emphasis on nervous system disorders and cancer. This review will update our understanding of PHF8 and promote the clinical transformation of its predictive and therapeutic value.
Collapse
Affiliation(s)
- Tingyu Fan
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang 421001, China; (T.F.); (G.H.)
| | - Jianlian Xie
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer, The Chinese University of Hong Kong, Hong Kong; (J.X.); (L.L.)
| | - Guo Huang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang 421001, China; (T.F.); (G.H.)
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen 518035, China
| | - Lili Li
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer, The Chinese University of Hong Kong, Hong Kong; (J.X.); (L.L.)
| | - Xi Zeng
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang 421001, China; (T.F.); (G.H.)
| | - Qian Tao
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer, The Chinese University of Hong Kong, Hong Kong; (J.X.); (L.L.)
| |
Collapse
|
2
|
Mishra J, Chakraborty S, Nandi P, Manna S, Baral T, Niharika, Roy A, Mishra P, Patra SK. Epigenetic regulation of androgen dependent and independent prostate cancer. Adv Cancer Res 2024; 161:223-320. [PMID: 39032951 DOI: 10.1016/bs.acr.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Prostate cancer is one of the most common malignancies among men worldwide. Besides genetic alterations, epigenetic modulations including DNA methylation, histone modifications and miRNA mediated alteration of gene expression are the key driving forces for the prostate tumor development and cancer progression. Aberrant expression and/or the activity of the epigenetic modifiers/enzymes, results in aberrant expression of genes involved in DNA repair, cell cycle regulation, cell adhesion, apoptosis, autophagy, tumor suppression and hormone response and thereby disease progression. Altered epigenome is associated with prostate cancer recurrence, progression, aggressiveness and transition from androgen-dependent to androgen-independent phenotype. These epigenetic modifications are reversible and various compounds/drugs targeting the epigenetic enzymes have been developed that are effective in cancer treatment. This chapter focuses on the epigenetic alterations in prostate cancer initiation and progression, listing different epigenetic biomarkers for diagnosis and prognosis of the disease and their potential as therapeutic targets. This chapter also summarizes different epigenetic drugs approved for prostate cancer therapy and the drugs available for clinical trials.
Collapse
Affiliation(s)
- Jagdish Mishra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Subhajit Chakraborty
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Piyasa Nandi
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Soumen Manna
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Tirthankar Baral
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Niharika
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Ankan Roy
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Prahallad Mishra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India.
| |
Collapse
|
3
|
Jian J, Wang X, Zhang J, Zhou C, Hou X, Huang Y, Hou J, Lin Y, Wei X. Molecular landscape for risk prediction and personalized therapeutics of castration-resistant prostate cancer: at a glance. Front Endocrinol (Lausanne) 2024; 15:1360430. [PMID: 38887275 PMCID: PMC11180744 DOI: 10.3389/fendo.2024.1360430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Prostate cancer (PCa) is commonly occurred with high incidence in men worldwide, and many patients will be eventually suffered from the dilemma of castration-resistance with the time of disease progression. Castration-resistant PCa (CRPC) is an advanced subtype of PCa with heterogeneous carcinogenesis, resulting in poor prognosis and difficulties in therapy. Currently, disorders in androgen receptor (AR)-related signaling are widely acknowledged as the leading cause of CRPC development, and some non-AR-based strategies are also proposed for CRPC clinical analyses. The initiation of CRPC is a consequence of abnormal interaction and regulation among molecules and pathways at multi-biological levels. In this study, CRPC-associated genes, RNAs, proteins, and metabolites were manually collected and integrated by a comprehensive literature review, and they were functionally classified and compared based on the role during CRPC evolution, i.e., drivers, suppressors, and biomarkers, etc. Finally, translational perspectives for data-driven and artificial intelligence-powered CRPC systems biology analysis were discussed to highlight the significance of novel molecule-based approaches for CRPC precision medicine and holistic healthcare.
Collapse
Affiliation(s)
- Jingang Jian
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin’an Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jun Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chenchao Zhou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaorui Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuhua Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuxin Lin
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Xuedong Wei
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
4
|
Tong D, Tang Y, Zhong P. The emerging roles of histone demethylases in cancers. Cancer Metastasis Rev 2024; 43:795-821. [PMID: 38227150 DOI: 10.1007/s10555-023-10160-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 12/05/2023] [Indexed: 01/17/2024]
Abstract
Modulation of histone methylation status is regarded as an important mechanism of epigenetic regulation and has substantial clinical potential for the therapy of diseases, including cancer and other disorders. The present study aimed to provide a comprehensive introduction to the enzymology of histone demethylases, as well as their cancerous roles, molecular mechanisms, therapeutic possibilities, and challenges for targeting them, in order to advance drug design for clinical therapy and highlight new insight into the mechanisms of these enzymes in cancer. A series of clinical trials have been performed to explore potential roles of histone demethylases in several cancer types. Numerous targeted inhibitors associated with immunotherapy, chemotherapy, radiotherapy, and targeted therapy have been used to exert anticancer functions. Future studies should evaluate the dynamic transformation of histone demethylases leading to carcinogenesis and explore individual therapy.
Collapse
Affiliation(s)
- Dali Tong
- Department of Urological Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China.
| | - Ying Tang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China.
| | - Peng Zhong
- Department of Pathology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China.
| |
Collapse
|
5
|
Qin L, Berk M, Chung YM, Cui D, Zhu Z, Chakraborty AA, Sharifi N. Chronic hypoxia stabilizes 3βHSD1 via autophagy suppression. Cell Rep 2024; 43:113575. [PMID: 38181788 PMCID: PMC10851248 DOI: 10.1016/j.celrep.2023.113575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 10/02/2023] [Accepted: 11/28/2023] [Indexed: 01/07/2024] Open
Abstract
Progression of prostate cancer depends on androgen receptor, which is usually activated by androgens. Therefore, a mainstay treatment is androgen deprivation therapy. Unfortunately, despite initial treatment response, resistance nearly always develops, and disease progresses to castration-resistant prostate cancer (CRPC), which remains driven by non-gonadal androgens synthesized in prostate cancer tissues. 3β-Hydroxysteroid dehydrogenase/Δ5-->4 isomerase 1 (3βHSD1) catalyzes the rate-limiting step in androgen synthesis. However, how 3βHSD1, especially the "adrenal-permissive" 3βHSD1(367T) that permits tumor synthesis of androgen from dehydroepiandrosterone (DHEA), is regulated at the protein level is not well understood. Here, we investigate how hypoxia regulates 3βHSD1(367T) protein levels. Our results show that, in vitro, hypoxia stabilizes 3βHSD1 protein by suppressing autophagy. Autophagy inhibition promotes 3βHSD1-dependent tumor progression. Hypoxia represses transcription of autophagy-related (ATG) genes by decreasing histone acetylation. Inhibiting deacetylase (HDAC) restores ATG gene transcription under hypoxia. Therefore, HDAC inhibition may be a therapeutic target for hypoxic tumor cells.
Collapse
Affiliation(s)
- Liang Qin
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Genitourinary Malignancies Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Michael Berk
- Genitourinary Malignancies Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yoon-Mi Chung
- Genitourinary Malignancies Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Di Cui
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Ziqi Zhu
- Genitourinary Malignancies Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Abhishek A Chakraborty
- Genitourinary Malignancies Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Nima Sharifi
- Genitourinary Malignancies Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
6
|
Tu X, Zhang J, Yuan W, Wu X, Xu Z, Qing C. Simvastatin Enhanced Anti-tumor Effects of Bevacizumab against Lung Adenocarcinoma A549 Cells via Abating HIF-1α-Wnt/β-Catenin Signaling Pathway. Anticancer Agents Med Chem 2023; 23:2083-2094. [PMID: 37587804 DOI: 10.2174/1871520623666230816090914] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/16/2023] [Accepted: 07/06/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Bevacizumab increased hypoxia-inducible factor (HIF-1α) expression attenuates its antitumor effect. Simvastatin can reduce the expression of HIF-1α to exert a tumor-suppressive effect in many in vitro experiments. Therefore, this study aimed to determine whether simvastatin could strengthen the anti-tumor activity of bevacizumab in lung adenocarcinoma. OBJECTIVE To determine whether simvastatin could strengthen the anti-tumor activity of bevacizumab in lung adenocarcinoma. METHODS The changes in the biological behavior of A549 cells treated with different drugs were determined through colony forming assay, Cell Counting Assay-8 (CCK-8), transwell assay, wound healing assay, and flow cytometry. The expressions of pathway-related factors HIF-1α and β-Catenin were determined via qRT-PCR and western blotting. The expressions of proliferation-related proteins, invasion-related proteins, and apoptosis-related proteins were detected by western blotting. In addition, a xenograft non-small cell lung cancer model in nude mice was used to explore in vivo tumor growth. RESULTS We found that simvastatin combined with bevacizumab synergistically suppressed the proliferation, migration, and invasion of A549 cells while promoting their apoptosis. As demonstrated by qRT-PCR and western blotting experiments, the bevacizumab group displayed a higher expression of pathway-related factors HIF-1α and β-Catenin than the control groups, however simvastatin group showed the opposite trend. Its combination with bevacizumab induced elevation of HIF-1α and β-catenin expressions. During in vivo experiments, simvastatin inhibited tumor growth, and in comparison, the inhibitory effects of its combination with bevacizumab were stronger. CONCLUSION Based on our findings, simvastatin may affect the biological responses of bevacizumab on A549 cells by restraining the HIF-1α-Wnt/β-catenin signaling pathway, thus representing a novel and effective combination therapy that can be potentially applied in a clinical therapy for lung adenocarcinoma.
Collapse
Affiliation(s)
- Xin Tu
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Chengdu Medical College, Chengdu Pidu District People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Jian Zhang
- Department of Gastroenterology, The Second People's Hospital of Yibin, Yibin, Sichuan, People's Republic of China
| | - Wei Yuan
- Department of Neurology, The Third Affiliated Hospital of Chengdu Medical College, Chengdu Pidu District People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Xia Wu
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Chengdu Medical College, Chengdu Pidu District People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Zhi Xu
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Chengdu Medical College, Chengdu Pidu District People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Cuo Qing
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Chengdu Medical College, Chengdu Pidu District People's Hospital, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
7
|
Shao P, Liu Q, Qi HH. KDM7 Demethylases: Regulation, Function and Therapeutic Targeting. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1433:167-184. [PMID: 37751140 DOI: 10.1007/978-3-031-38176-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
It was more than a decade ago that PHF8, KDM7A/JHDM1D and PHF2 were first proposed to be a histone demethylase family and were named as KDM7 (lysine demethylase) family. Since then, knowledge of their demethylation activities, roles as co-regulators of transcription and roles in development and diseases such as cancer has been steadily growing. The demethylation activities of PHF8 and KDM7A toward various methylated histones including H3K9me2/1, H3K27me2 and H4K20me1 have been identified and proven in various cell types. In contrast, PHF2, due to a mutation of a key residue in an iron-binding domain, demethylates H3K9me2 upon PKA-mediated phosphorylation. Interestingly, it was reported that PHF2 possesses an unusual H4K20me3 demethylation activity, which was not observed for PHF8 and KDM7A. PHF8 has been most extensively studied with respect to its roles in development and oncogenesis, revealing that it contributes to regulation of the cell cycle, cell viability and cell migration. Moreover, accumulating lines of evidence demonstrated that the KDM7 family members are subjected to post-transcriptional and post-translational regulations, leading to a higher horizon for evaluating their actual protein expression and functions in development and cancer. This chapter provides a general view of the current understanding of the regulation and functions of the KDM7 family and discusses their potential as therapeutic targets in cancer as well as perspectives for further studies.
Collapse
Affiliation(s)
- Peng Shao
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, 51 Newton Road, Iowa City, IA, 52242, USA
| | - Qi Liu
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, 51 Newton Road, Iowa City, IA, 52242, USA
| | - Hank Heng Qi
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, 51 Newton Road, Iowa City, IA, 52242, USA.
| |
Collapse
|
8
|
Sawada T, Kanemoto Y, Kurokawa T, Kato S. The epigenetic function of androgen receptor in prostate cancer progression. Front Cell Dev Biol 2023; 11:1083486. [PMID: 37025180 PMCID: PMC10070878 DOI: 10.3389/fcell.2023.1083486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
Androgen and androgen deprivation (castration) therapies, including androgen receptor antagonists, are clinically used to treat patients with prostate cancer. However, most hormone-dependent prostate cancer patients progress into a malignant state with loss of hormone-dependency, known as castration (drug)-resistant prostate cancer (CRPC), after prolong androgen-based treatments. Even in the CRPC state with irreversible malignancy, androgen receptor (AR) expression is detectable. An epigenetic transition to CRPC induced by the action of AR-mediated androgen could be speculated in the patients with prostate cancer. Androgen receptors belongs to the nuclear receptor superfamily with 48 members in humans, and acts as a ligand-dependent transcriptional factor, leading to local chromatin reorganization for ligand-dependent gene regulation. In this review, we discussed the transcriptional/epigenetic regulatory functions of AR, with emphasis on the clinical applications of AR ligands, AR protein co-regulators, and AR RNA coregulator (enhancer RNA), especially in chromatin reorganization, in patients with prostate cancer.
Collapse
Affiliation(s)
- Takahiro Sawada
- Graduate School of Life Science and Engineering, Iryo Sosei University, Fukushima, Japan
- Research Institute of Innovative Medicine, Tokiwa Foundation, Fukushima, Japan
| | - Yoshiaki Kanemoto
- Graduate School of Life Science and Engineering, Iryo Sosei University, Fukushima, Japan
- Research Institute of Innovative Medicine, Tokiwa Foundation, Fukushima, Japan
| | - Tomohiro Kurokawa
- Graduate School of Life Science and Engineering, Iryo Sosei University, Fukushima, Japan
- Research Institute of Innovative Medicine, Tokiwa Foundation, Fukushima, Japan
- School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Shigeaki Kato
- Graduate School of Life Science and Engineering, Iryo Sosei University, Fukushima, Japan
- Research Institute of Innovative Medicine, Tokiwa Foundation, Fukushima, Japan
- School of Medicine, Fukushima Medical University, Fukushima, Japan
- *Correspondence: Shigeaki Kato,
| |
Collapse
|
9
|
NEDD4L represses prostate cancer cell proliferation via modulating PHF8 through the ubiquitin-proteasome pathway. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:243-255. [PMID: 36136271 DOI: 10.1007/s12094-022-02933-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/22/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE Prostate cancer (PC) is a heterogeneous malignancy that greatly threatens man's health. E3 ubiquitin-protein ligase neural precursor cell expressed developmentally downregulated 4-like (NEDD4L) imparts an regulatory role in various malignancies. This study focused on the modulatory mechanism of NEDD4L in proliferation of prostate cancer cells (PCCs) via regulating histone demethylase plant homeodomain finger protein 8 (PHF8/KDM7B) through the ubiquitin-proteasome system. METHODS The expression levels of NEDD4L, PHF8, H3 lysine 9 dimethylation (H3K9me2) and activating transcription factor 2 (ATF2) in PC tissues and cell lines were detected via real-time quantitative polymerase chain reaction and Western blotting. After transfection of pcDNA3.1-NEDD4L, pcDNA3.1-PHF8, and pcDNA3.1-ATF2 into PCCs, cell proliferation was assessed via the cell counting kit-8 and 5-ethynyl-2'-deoxyuridine assays. Interaction between NEDD4L and PHF8 was identified via the protein immunoprecipitation. The ubiquitination level of PHF8 was determined via the ubiquitination detection. The enrichments of H3K9me2 and PHF8 in the ATF2 promotor region were detected via the chromatin-immunoprecipitation assay. RESULTS PHF8 and ATF2 were highly expressed while NEDD4L was poorly expressed in PC tissues and cells. NEDD4L overexpression reduced proliferation of PCCs. NEDD4Linduced degradation of PHF8 via ubiquitination. PHF8 limited the enrichment of H3K9me2 in the ATF2 promotor region and enhanced ATF2 transcription. Upregulation of PHF8 or ATF2 abolished the inhibitory role of NEDD4L in proliferation of PCCs. CONCLUSION NEDD4L facilitated degradation of PHF8 to limit ATF2 transcription, thereby suppressing proliferation of PCCs.
Collapse
|
10
|
Diao W, Zheng J, Li Y, Wang J, Xu S. Targeting histone demethylases as a potential cancer therapy (Review). Int J Oncol 2022; 61:103. [PMID: 35801593 DOI: 10.3892/ijo.2022.5393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/15/2022] [Indexed: 11/06/2022] Open
Abstract
Post‑translational modifications of histones by histone demethylases have an important role in the regulation of gene transcription and are implicated in cancers. Recently, the family of lysine (K)‑specific demethylase (KDM) proteins, referring to histone demethylases that dynamically regulate histone methylation, were indicated to be involved in various pathways related to cancer development. To date, numerous studies have been conducted to explore the effects of KDMs on cancer growth, metastasis and drug resistance, and a majority of KDMs have been indicated to be oncogenes in both leukemia and solid tumors. In addition, certain KDM inhibitors have been developed and have become the subject of clinical trials to explore their safety and efficacy in cancer therapy. However, most of them focus on hematopoietic malignancy. This review summarizes the effects of KDMs on tumor growth, drug resistance and the current status of KDM inhibitors in clinical trials.
Collapse
Affiliation(s)
- Wenfei Diao
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Jiabin Zheng
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Yong Li
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Junjiang Wang
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Songhui Xu
- Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
11
|
Xie Y, Ning S, Hu J. Molecular mechanisms of neuroendocrine differentiation in prostate cancer progression. J Cancer Res Clin Oncol 2022; 148:1813-1823. [PMID: 35633416 PMCID: PMC9189092 DOI: 10.1007/s00432-022-04061-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/09/2022] [Indexed: 12/03/2022]
Abstract
Background Rapid evolution of the therapeutic management of prostate cancer, especially in in second-generation androgen inhibitors, has increased the opportunity of transformation from prostate cancer (PCa) to neuroendocrine prostate cancer (NEPC). NEPC still lacks effective diagnostic and therapeutic interventions. Researches into the molecular characteristics of neuroendocrine differentiation is undoubtedly crucial to the discovery of new target genes for accurate diagnostic and therapeutic targets. Purpose In this review, we focus on the relevant genes and molecular mechanisms that have contributed to the transformation in the progression of PCa and discuss the potential targeted molecule that might improve diagnostic accuracy and therapeutic effectiveness. Methods The relevant literatures from PubMed have been reviewed for this article. Conclusion Several molecular characteristics influence the progression of neuroendocrine differentiation of prostate cancer which will provide a novel sight for accurate diagnosis and target therapeutic intervention for patients with NEPC.
Collapse
Affiliation(s)
- Yuchen Xie
- Affiliated Renmin Hospital of Jiangsu University, Zhenjiang First People's Hospital, Zhenjiang, 212002, China
| | - Songyi Ning
- Jiangsu University, Zhenjiang, 212013, China
| | - Jianpeng Hu
- Affiliated Renmin Hospital of Jiangsu University, Zhenjiang First People's Hospital, Zhenjiang, 212002, China.
| |
Collapse
|
12
|
López J, Añazco-Guenkova AM, Monteagudo-García Ó, Blanco S. Epigenetic and Epitranscriptomic Control in Prostate Cancer. Genes (Basel) 2022; 13:genes13020378. [PMID: 35205419 PMCID: PMC8872343 DOI: 10.3390/genes13020378] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/12/2022] [Accepted: 02/16/2022] [Indexed: 12/19/2022] Open
Abstract
The initiation of prostate cancer has been long associated with DNA copy-number alterations, the loss of specific chromosomal regions and gene fusions, and driver mutations, especially those of the Androgen Receptor. Non-mutational events, particularly DNA and RNA epigenetic dysregulation, are emerging as key players in tumorigenesis. In this review we summarize the molecular changes linked to epigenetic and epitranscriptomic dysregulation in prostate cancer and the role that alterations to DNA and RNA modifications play in the initiation and progression of prostate cancer.
Collapse
Affiliation(s)
- Judith López
- Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)—University of Salamanca, 37007 Salamanca, Spain; (J.L.); (A.M.A.-G.); (Ó.M.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Ana M. Añazco-Guenkova
- Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)—University of Salamanca, 37007 Salamanca, Spain; (J.L.); (A.M.A.-G.); (Ó.M.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Óscar Monteagudo-García
- Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)—University of Salamanca, 37007 Salamanca, Spain; (J.L.); (A.M.A.-G.); (Ó.M.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Sandra Blanco
- Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)—University of Salamanca, 37007 Salamanca, Spain; (J.L.); (A.M.A.-G.); (Ó.M.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Correspondence:
| |
Collapse
|
13
|
Moubarak RS, de Pablos-Aragoneses A, Ortiz-Barahona V, Gong Y, Gowen M, Dolgalev I, Shadaloey SAA, Argibay D, Karz A, Von Itter R, Vega-Sáenz de Miera EC, Sokolova E, Darvishian F, Tsirigos A, Osman I, Hernando E. The histone demethylase PHF8 regulates TGFβ signaling and promotes melanoma metastasis. SCIENCE ADVANCES 2022; 8:eabi7127. [PMID: 35179962 PMCID: PMC8856617 DOI: 10.1126/sciadv.abi7127] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 12/14/2021] [Indexed: 05/17/2023]
Abstract
The contribution of epigenetic dysregulation to metastasis remains understudied. Through a meta-analysis of gene expression datasets followed by a mini-screen, we identified Plant Homeodomain Finger protein 8 (PHF8), a histone demethylase of the Jumonji C protein family, as a previously unidentified prometastatic gene in melanoma. Loss- and gain-of-function approaches demonstrate that PHF8 promotes cell invasion without affecting proliferation in vitro and increases dissemination but not subcutaneous tumor growth in vivo, thus supporting its specific contribution to the acquisition of metastatic potential. PHF8 requires its histone demethylase activity to enhance melanoma cell invasion. Transcriptomic and epigenomic analyses revealed that PHF8 orchestrates a molecular program that directly controls the TGFβ signaling pathway and, as a consequence, melanoma invasion and metastasis. Our findings bring a mechanistic understanding of epigenetic regulation of metastatic fitness in cancer, which may pave the way for improved therapeutic interventions.
Collapse
Affiliation(s)
- Rana S. Moubarak
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | | | | | - Yixiao Gong
- Applied Bioinformatics Laboratories, NYU School of Medicine, NY 10016, USA
| | - Michael Gowen
- NYU School of Medicine Institute for Computational Medicine, New York, NY 10016, USA
| | - Igor Dolgalev
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Applied Bioinformatics Laboratories, NYU School of Medicine, NY 10016, USA
| | - Sorin A. A. Shadaloey
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
| | - Diana Argibay
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
| | - Alcida Karz
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
| | - Richard Von Itter
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
| | | | - Elena Sokolova
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
| | - Farbod Darvishian
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
| | - Aristotelis Tsirigos
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Applied Bioinformatics Laboratories, NYU School of Medicine, NY 10016, USA
- NYU School of Medicine Institute for Computational Medicine, New York, NY 10016, USA
| | - Iman Osman
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
- Ronald O. Perelman Department of Dermatology, NYU School of Medicine, New York, NY 10016, USA
| | - Eva Hernando
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| |
Collapse
|
14
|
Merkens L, Sailer V, Lessel D, Janzen E, Greimeier S, Kirfel J, Perner S, Pantel K, Werner S, von Amsberg G. Aggressive variants of prostate cancer: underlying mechanisms of neuroendocrine transdifferentiation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:46. [PMID: 35109899 PMCID: PMC8808994 DOI: 10.1186/s13046-022-02255-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/13/2022] [Indexed: 12/14/2022]
Abstract
Prostate cancer is a hormone-driven disease and its tumor cell growth highly relies on increased androgen receptor (AR) signaling. Therefore, targeted therapy directed against androgen synthesis or AR activation is broadly used and continually improved. However, a subset of patients eventually progresses to castration-resistant disease. To date, various mechanisms of resistance have been identified including the development of AR-independent aggressive variant prostate cancer based on neuroendocrine transdifferentiation (NED). Here, we review the highly complex processes contributing to NED. Genetic, epigenetic, transcriptional aberrations and posttranscriptional modifications are highlighted and the potential interplay of the different factors is discussed. Background Aggressive variant prostate cancer (AVPC) with traits of neuroendocrine differentiation emerges in a rising number of patients in recent years. Among others, advanced therapies targeting the androgen receptor axis have been considered causative for this development. Cell growth of AVPC often occurs completely independent of the androgen receptor signal transduction pathway and cells have mostly lost the typical cellular features of prostate adenocarcinoma. This complicates both diagnosis and treatment of this very aggressive disease. We believe that a deeper understanding of the complex molecular pathological mechanisms contributing to transdifferentiation will help to improve diagnostic procedures and develop effective treatment strategies. Indeed, in recent years, many scientists have made important contributions to unravel possible causes and mechanisms in the context of neuroendocrine transdifferentiation. However, the complexity of the diverse molecular pathways has not been captured completely, yet. This narrative review comprehensively highlights the individual steps of neuroendocrine transdifferentiation and makes an important contribution in bringing together the results found so far.
Collapse
Affiliation(s)
- Lina Merkens
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| | - Verena Sailer
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Campus Luebeck, Ratzeburger Allee 160, 23538, Luebeck, Germany
| | - Davor Lessel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Ella Janzen
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Sarah Greimeier
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Jutta Kirfel
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Campus Luebeck, Ratzeburger Allee 160, 23538, Luebeck, Germany
| | - Sven Perner
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Campus Luebeck, Ratzeburger Allee 160, 23538, Luebeck, Germany.,Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.,European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Stefan Werner
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.,Mildred Scheel Cancer Career Center Hamburg HaTRiCs4, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gunhild von Amsberg
- Department of Hematology and Oncology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.,Martini-Klinik, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| |
Collapse
|
15
|
Abstract
Hypoxia is defined as a cellular stress condition caused by a decrease in oxygen below physiologically normal levels. Cells in the core of a rapidly growing solid tumor are faced with the challenge of inadequate supply of oxygen through the blood, owing to improper vasculature inside the tumor. This hypoxic microenvironment inside the tumor initiates a gene expression program that alters numerous signaling pathways, allowing the cancer cell to eventually evade adverse conditions and attain a more aggressive phenotype. A multitude of studies covering diverse aspects of gene regulation has tried to uncover the mechanisms involved in hypoxia-induced tumorigenesis. The role of epigenetics in executing widespread and dynamic changes in gene expression under hypoxia has been gaining an increasing amount of support in recent years. This chapter discusses, in detail, various epigenetic mechanisms driving the cellular response to hypoxia in cancer.
Collapse
Affiliation(s)
- Deepak Pant
- Epigenetics and RNA Processing Lab (ERPL), Indian Institute of Science Education and Research Bhopal, Bhopal, India
| | - Srinivas Abhishek Mutnuru
- Epigenetics and RNA Processing Lab (ERPL), Indian Institute of Science Education and Research Bhopal, Bhopal, India
| | - Sanjeev Shukla
- Epigenetics and RNA Processing Lab (ERPL), Indian Institute of Science Education and Research Bhopal, Bhopal, India.
| |
Collapse
|
16
|
Deng T, Xiao Y, Dai Y, Xie L, Li X. Roles of Key Epigenetic Regulators in the Gene Transcription and Progression of Prostate Cancer. Front Mol Biosci 2021; 8:743376. [PMID: 34977151 PMCID: PMC8714908 DOI: 10.3389/fmolb.2021.743376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 11/25/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is a top-incidence malignancy, and the second most common cause of death amongst American men and the fifth leading cause of cancer death in men around the world. Androgen receptor (AR), the key transcription factor, is critical for the progression of PCa by regulating a series of target genes by androgen stimulation. A number of co-regulators of AR, including co-activators or co-repressors, have been implicated in AR-mediated gene transcription and PCa progression. Epigenetic regulators, by modifying chromatin integrity and accessibility for transcription regulation without altering DNA sequences, influence the transcriptional activity of AR and further regulate the gene expression of AR target genes in determining cell fate, PCa progression and therapeutic response. In this review, we summarized the structural interaction of AR and epigenetic regulators including histone or DNA methylation, histone acetylation or non-coding RNA, and functional synergy in PCa progression. Importantly, epigenetic regulators have been validated as diagnostic markers and therapeutic targets. A series of epigenetic target drugs have been developed, and have demonstrated the potential to treat PCa alone or in combination with antiandrogens.
Collapse
Affiliation(s)
- Tanggang Deng
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yugang Xiao
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yi Dai
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lin Xie
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiong Li
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
17
|
Alcantara-Zapata DE, Llanos AJ, Nazzal C. High altitude exposure affects male reproductive parameters: Could it also affect the prostate?†. Biol Reprod 2021; 106:385-396. [PMID: 34725677 DOI: 10.1093/biolre/ioab205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/26/2021] [Indexed: 11/14/2022] Open
Abstract
Living at high altitudes and living with prostatic illness are two different conditions closely related to a hypoxic environment. People at high altitudes exposed to acute, chronic, or intermittent hypobaric hypoxia turn on several mechanisms at the system, cellular and molecular level to cope with oxygen atmosphere scarcity maintaining the oxygen homeostasis. This exposure affects the whole organism and function of many systems, such as cardiovascular, respiratory, and reproductive. On the other hand, malignant prostate is related to the scarcity of oxygen in the tissue microenvironment due to its low availability and high consumption due to the swift cell proliferation rates. Based on the literature, this similarity in the oxygen scarcity suggests that hypobaric hypoxia, and other common factors between these two conditions, could be involved in the aggravation of the pathological prostatic status. However, there is still a lack of evidence in the association of this disease in males at high altitudes. This review aims to examine the possible mechanisms that hypobaric hypoxia might negatively add to the pathological prostate function in males who live and work at high altitudes. More profound investigations of hypobaric hypoxia's direct action on the prostate could help understand this exposure's effect and prevent worse prostate illness impact in males at high altitudes.
Collapse
Affiliation(s)
| | - Aníbal J Llanos
- Laboratorio de Fisiología y Fisiopatología del Desarrollo, Programa de Fisiopatología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Centro Internacional de Estudios Andinos (INCAS), Universidad de Chile, Santiago, Chile
| | - Carolina Nazzal
- Department of Epidemiology. School of Public Health. Faculty of Medicine. University of Chile
| |
Collapse
|
18
|
Casillas AL, Chauhan SS, Toth RK, Sainz AG, Clements AN, Jensen CC, Langlais PR, Miranti CK, Cress AE, Warfel NA. Direct phosphorylation and stabilization of HIF-1α by PIM1 kinase drives angiogenesis in solid tumors. Oncogene 2021; 40:5142-5152. [PMID: 34211090 PMCID: PMC8364516 DOI: 10.1038/s41388-021-01915-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 06/09/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022]
Abstract
Angiogenesis is essential for the sustained growth of solid tumors. Hypoxia-inducible factor 1 (HIF-1) is a master regulator of angiogenesis and constitutive activation of HIF-1 is frequently observed in human cancers. Therefore, understanding the mechanisms governing the activation of HIF-1 is critical for successful therapeutic targeting of tumor angiogenesis. Herein, we establish a new regulatory mechanism responsible for the constitutive activation of HIF-1α in cancer, irrespective of oxygen tension. PIM1 kinase directly phosphorylates HIF-1α at threonine 455, a previously uncharacterized site within its oxygen-dependent degradation domain. This phosphorylation event disrupts the ability of prolyl hydroxylases to bind and hydroxylate HIF-1α, interrupting its canonical degradation pathway and promoting constitutive transcription of HIF-1 target genes. Moreover, phosphorylation of the analogous site in HIF-2α (S435) stabilizes the protein through the same mechanism, indicating post-translational modification within the oxygen-dependent degradation domain as a mechanism of regulating the HIF-α subunits. In vitro and in vivo models demonstrate that expression of PIM1 is sufficient to stabilize HIF-1α and HIF-2α in normoxia and stimulate angiogenesis in a HIF-1-dependent manner. CRISPR mutants of HIF-1α (Thr455D) promoted increased tumor growth, proliferation, and angiogenesis. Moreover, HIF-1α-T455D xenograft tumors were refractory to the anti-angiogenic and cytotoxic effects of PIM inhibitors. These data identify a new signaling axis responsible for hypoxia-independent activation of HIF-1 and expand our understanding of the tumorigenic role of PIM1 in solid tumors.
Collapse
Affiliation(s)
- Andrea L Casillas
- Cancer Biology Graduate Interdisciplinary Program, The University of Arizona, Tucson, AZ, USA
| | | | - Rachel K Toth
- The University of Arizona Cancer Center, Tucson, AZ, USA
| | - Alva G Sainz
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Amber N Clements
- Cancer Biology Graduate Interdisciplinary Program, The University of Arizona, Tucson, AZ, USA
| | - Corbin C Jensen
- Cancer Biology Graduate Interdisciplinary Program, The University of Arizona, Tucson, AZ, USA
| | - Paul R Langlais
- Department of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Cindy K Miranti
- The University of Arizona Cancer Center, Tucson, AZ, USA
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, USA
| | - Anne E Cress
- The University of Arizona Cancer Center, Tucson, AZ, USA
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, USA
| | - Noel A Warfel
- The University of Arizona Cancer Center, Tucson, AZ, USA.
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
19
|
Pacheco MB, Camilo V, Henrique R, Jerónimo C. Epigenetic Editing in Prostate Cancer: Challenges and Opportunities. Epigenetics 2021; 17:564-588. [PMID: 34130596 DOI: 10.1080/15592294.2021.1939477] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Epigenome editing consists of fusing a predesigned DNA recognition unit to the catalytic domain of a chromatin modifying enzyme leading to the introduction or removal of an epigenetic mark at a specific locus. These platforms enabled the study of the mechanisms and roles of epigenetic changes in several research domains such as those addressing pathogenesis and progression of cancer. Despite the continued efforts required to overcome some limitations, which include specificity, off-target effects, efficacy, and longevity, these tools have been rapidly progressing and improving.Since prostate cancer is characterized by multiple genetic and epigenetic alterations that affect different signalling pathways, epigenetic editing constitutes a promising strategy to hamper cancer progression. Therefore, by modulating chromatin structure through epigenome editing, its conformation might be better understood and events that drive prostate carcinogenesis might be further unveiled.This review describes the different epigenome engineering tools, their mechanisms concerning gene's expression and regulation, highlighting the challenges and opportunities concerning prostate cancer research.
Collapse
Affiliation(s)
- Mariana Brütt Pacheco
- Cancer Biology and Epigenetics Group, Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, Porto, Portugal
| | - Vânia Camilo
- Cancer Biology and Epigenetics Group, Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), R. DR. António Bernardino De Almeida, Porto, Portugal.,Department of Pathology and Molecular Immunology, School of Medicine & Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, Porto, Portugal.,Department of Pathology and Molecular Immunology, School of Medicine & Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, Porto, Portugal
| |
Collapse
|
20
|
Liu Q, Pang J, Wang L, Huang Z, Xu J, Yang X, Xie Q, Huang Y, Tang T, Tong D, Liu G, Wang L, Zhang D, Ma Q, Xiao H, Lan W, Qin J, Jiang J. Histone demethylase PHF8 drives neuroendocrine prostate cancer progression by epigenetically upregulating FOXA2. J Pathol 2021; 253:106-118. [PMID: 33009820 PMCID: PMC7756255 DOI: 10.1002/path.5557] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/08/2020] [Accepted: 09/28/2020] [Indexed: 01/20/2023]
Abstract
Neuroendocrine prostate cancer (NEPC) is a more aggressive subtype of castration-resistant prostate cancer (CRPC). Although it is well established that PHF8 can enhance prostate cancer cell proliferation, whether PHF8 is involved in prostate cancer initiation and progression is relatively unclear. By comparing the transgenic adenocarcinoma of the mouse prostate (TRAMP) mice with or without Phf8 knockout, we systemically examined the role of PHF8 in prostate cancer development. We found that PHF8 plays a minimum role in initiation and progression of adenocarcinoma. However, PHF8 is essential for NEPC because not only is PHF8 highly expressed in NEPC but also animals without Phf8 failed to develop NEPC. Mechanistically, PHF8 transcriptionally upregulates FOXA2 by demethylating and removing the repressive histone markers on the promoter region of the FOXA2 gene, and the upregulated FOXA2 subsequently regulates the expression of genes involved in NEPC development. Since both PHF8 and FOXA2 are highly expressed in NEPC tissues from patients or patient-derived xenografts, the levels of PHF8 and FOXA2 can either individually or in combination serve as NEPC biomarkers and targeting either PHF8 or FOXA2 could be potential therapeutic strategies for NEPC treatment. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
MESH Headings
- Adenocarcinoma/enzymology
- Adenocarcinoma/genetics
- Adenocarcinoma/secondary
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Neuroendocrine/enzymology
- Carcinoma, Neuroendocrine/genetics
- Carcinoma, Neuroendocrine/secondary
- Cell Movement
- Cell Proliferation
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic
- Hepatocyte Nuclear Factor 3-beta/genetics
- Hepatocyte Nuclear Factor 3-beta/metabolism
- Histone Demethylases/genetics
- Histone Demethylases/metabolism
- Humans
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Nude
- PC-3 Cells
- Prostatic Neoplasms/enzymology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/pathology
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic
- Up-Regulation
- Mice
Collapse
Affiliation(s)
- Qiuli Liu
- Department of Urology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| | - Jian Pang
- Department of Urology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| | - Lin‐ang Wang
- Department of Urology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| | - Zhuowei Huang
- Department of Urology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| | - Jing Xu
- Department of Urology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| | - Xingxia Yang
- Department of Urology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| | - Qiubo Xie
- Department of Urology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| | - Yiqiang Huang
- Department of Urology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| | - Tang Tang
- Department of Urology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| | - Dali Tong
- Department of Urology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| | - Gaolei Liu
- Department of Urology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| | - Luofu Wang
- Department of Urology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| | - Dianzheng Zhang
- Department of Bio‐Medical SciencesPhiladelphia College of Osteopathic MedicinePhiladelphiaPAUSA
| | - Qiang Ma
- Department of Pathology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| | - Hualiang Xiao
- Department of Pathology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| | - Weihua Lan
- Department of Urology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| | - Jun Qin
- Department of Urology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Nutrition and Health SciencesChinese Academy of Sciences, University of Chinese Academy of SciencesShanghaiPR China
| | - Jun Jiang
- Department of Urology, Institute of Surgery ResearchDaping Hospital, Army Medical UniversityChongqingPR China
| |
Collapse
|
21
|
VanderVeen BN, Murphy EA, Carson JA. The Impact of Immune Cells on the Skeletal Muscle Microenvironment During Cancer Cachexia. Front Physiol 2020; 11:1037. [PMID: 32982782 PMCID: PMC7489038 DOI: 10.3389/fphys.2020.01037] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/28/2020] [Indexed: 12/22/2022] Open
Abstract
Progressive weight loss combined with skeletal muscle atrophy, termed cachexia, is a common comorbidity associated with cancer that results in adverse consequences for the patient related to decreased chemotherapy responsiveness and increased mortality. Cachexia's complexity has provided a barrier for developing successful therapies to prevent or treat the condition, since a large number of systemic disruptions that can regulate muscle mass are often present. Furthermore, considerable effort has focused on investigating how tumor derived factors and inflammatory mediators directly signal skeletal muscle to disrupt protein turnover regulation. Currently, there is developing appreciation for understanding how cancer alters skeletal muscle's complex microenvironment and the tightly regulated interactions between multiple cell types. Skeletal muscle microenvironment interactions have established functions in muscle response to regeneration from injury, growth, aging, overload-induced hypertrophy, and exercise. This review explores the growing body of evidence for immune cell modulation of the skeletal muscle microenvironment during cancer-induced muscle wasting. Emphasis is placed on the regulatory network that integrates physiological responses between immune cells with other muscle cell types including satellite cells, fibroblast cells, and endothelial cells to regulate myofiber size and plasticity. The overall goal of this review is to provide an understanding of how different cell types that constitute the muscle microenvironment and their signaling mediators contribute to cancer and chemotherapy-induced muscle wasting.
Collapse
Affiliation(s)
- Brandon N. VanderVeen
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
- AcePre, LLC, Columbia, SC, United States
| | - E. Angela Murphy
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
- AcePre, LLC, Columbia, SC, United States
| | - James A. Carson
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
22
|
Kim I, Park JW. Hypoxia-driven epigenetic regulation in cancer progression: A focus on histone methylation and its modifying enzymes. Cancer Lett 2020; 489:41-49. [PMID: 32522693 DOI: 10.1016/j.canlet.2020.05.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/28/2020] [Accepted: 05/20/2020] [Indexed: 02/08/2023]
Abstract
The mechanism underlying hypoxia-driven chromatin remodeling is a long-lasting question. For the last two decades, this question has been resolved in part. It is now widely agreed that hypoxia dynamically changes the methylation status of histones to control gene expression. Hypoxia-inducible factor (HIF) plays a central role in cellular responses to hypoxia through transcriptional activation of numerous genes. At least in part, the hypoxic regulation of histone methylation is attributed to the HIF-mediated expression of histone modifying enzymes. Protein hydroxylation and histone demethylation have emerged as the oxygen sensing processes because they are catalyzed by a family of 2-oxoglutarate (2OG)-dependent dioxygenases whose activities depend upon the ambient oxygen level. Recently, it has been extensively investigated that the 2OG dioxygenases oxygen-dependently regulate histone methylation. Nowadays, the hypoxic change in the histone methylation status is regarded as an important event to drive malignant behaviors of cancer cells. In this review, we introduced and summarized the cellular processes that govern hypoxia-driven regulation of histone methylation in the context of cancer biology. We also discussed the emerging roles of histone methyltransferases and demethylases in epigenetic response to hypoxia.
Collapse
Affiliation(s)
- Iljin Kim
- Department of Pharmacology, Cancer Research Institute, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jong-Wan Park
- Department of Pharmacology, Cancer Research Institute, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
23
|
Tran MGB, Bibby BAS, Yang L, Lo F, Warren AY, Shukla D, Osborne M, Hadfield J, Carroll T, Stark R, Scott H, Ramos-Montoya A, Massie C, Maxwell P, West CML, Mills IG, Neal DE. Independence of HIF1a and androgen signaling pathways in prostate cancer. BMC Cancer 2020; 20:469. [PMID: 32450824 PMCID: PMC7249645 DOI: 10.1186/s12885-020-06890-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/22/2020] [Indexed: 01/27/2023] Open
Abstract
Background Therapeutic targeting of the androgen signaling pathway is a mainstay treatment for prostate cancer. Although initially effective, resistance to androgen targeted therapies develops followed by disease progression to castrate-resistant prostate cancer (CRPC). Hypoxia and HIF1a have been implicated in the development of resistance to androgen targeted therapies and progression to CRCP. The interplay between the androgen and hypoxia/HIF1a signaling axes was investigated. Methods In vitro stable expression of HIF1a was established in the LNCaP cell line by physiological induction or retroviral transduction. Tumor xenografts with stable expression of HIF1a were established in castrated and non-castrated mouse models. Gene expression analysis identified transcriptional changes in response to androgen treatment, hypoxia and HIF1a. The binding sites of the AR and HIF transcription factors were identified using ChIP-seq. Results Androgen and HIF1a signaling promoted proliferation in vitro and enhanced tumor growth in vivo. The stable expression of HIF1a in vivo restored tumor growth in the absence of endogenous androgens. Hypoxia reduced AR binding sites whereas HIF binding sites were increased with androgen treatment under hypoxia. Gene expression analysis identified seven genes that were upregulated both by AR and HIF1a, of which six were prognostic. Conclusions The oncogenic AR, hypoxia and HIF1a pathways support prostate cancer development through independent signaling pathways and transcriptomic profiles. AR and hypoxia/HIF1a signaling pathways independently promote prostate cancer progression and therapeutic targeting of both pathways simultaneously is warranted.
Collapse
Affiliation(s)
- Maxine G B Tran
- Uro-oncology Research Group, Cancer Research UK Cambridge Institute, Cambridge, CB02 0RE, UK.,UCL division of Surgery and Interventional Science, Royal Free Hospital, Pond Street, London, NW3 2QG, UK
| | - Becky A S Bibby
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital NHS Trust, M20 4BX, Manchester, UK
| | - Lingjian Yang
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital NHS Trust, M20 4BX, Manchester, UK
| | - Franklin Lo
- Uro-oncology Research Group, Cancer Research UK Cambridge Institute, Cambridge, CB02 0RE, UK
| | - Anne Y Warren
- Department of Pathology, Addenbrooke's Cambridge University Hospital, Cambridge, UK
| | - Deepa Shukla
- Division of Medicine, University College London, London, UK
| | - Michelle Osborne
- Uro-oncology Research Group, Cancer Research UK Cambridge Institute, Cambridge, CB02 0RE, UK
| | - James Hadfield
- Uro-oncology Research Group, Cancer Research UK Cambridge Institute, Cambridge, CB02 0RE, UK
| | - Thomas Carroll
- Uro-oncology Research Group, Cancer Research UK Cambridge Institute, Cambridge, CB02 0RE, UK
| | - Rory Stark
- Uro-oncology Research Group, Cancer Research UK Cambridge Institute, Cambridge, CB02 0RE, UK
| | - Helen Scott
- Uro-oncology Research Group, Cancer Research UK Cambridge Institute, Cambridge, CB02 0RE, UK
| | - Antonio Ramos-Montoya
- Uro-oncology Research Group, Cancer Research UK Cambridge Institute, Cambridge, CB02 0RE, UK
| | - Charlie Massie
- Uro-oncology Research Group, Cancer Research UK Cambridge Institute, Cambridge, CB02 0RE, UK.,Department of Oncology, University of Cambridge, London, CB2 0XZ, UK
| | - Patrick Maxwell
- Cambridge Institute of Medical Research, Cambridge Biomedical Campus, Cambridge, CB2 0SP, UK
| | - Catharine M L West
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital NHS Trust, M20 4BX, Manchester, UK.,Manchester Biomedical Research Centre, University of Manchester, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Ian G Mills
- Patrick G Johnston Centre for Cancer Research and Cell Biology, Queens University Belfast, Belfast, BT9 7AE, UK. .,Nuffield Department of Surgical Sciences, University of Oxford, Oxford, OX3 9DU, UK.
| | - David E Neal
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, OX3 9DU, UK.,Academic Urology Group, University of Cambridge, Cambridge, UK
| |
Collapse
|
24
|
Saraç H, Morova T, Pires E, McCullagh J, Kaplan A, Cingöz A, Bagci-Onder T, Önder T, Kawamura A, Lack NA. Systematic characterization of chromatin modifying enzymes identifies KDM3B as a critical regulator in castration resistant prostate cancer. Oncogene 2020; 39:2187-2201. [PMID: 31822799 PMCID: PMC7056651 DOI: 10.1038/s41388-019-1116-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 11/05/2019] [Accepted: 11/11/2019] [Indexed: 12/28/2022]
Abstract
Androgen deprivation therapy (ADT) is the standard care for prostate cancer (PCa) patients who fail surgery or radiotherapy. While initially effective, the cancer almost always recurs as a more aggressive castration resistant prostate cancer (CRPC). Previous studies have demonstrated that chromatin modifying enzymes can play a critical role in the conversion to CRPC. However, only a handful of these potential pharmacological targets have been tested. Therefore, in this study, we conducted a focused shRNA screen of chromatin modifying enzymes previously shown to be involved in cellular differentiation. We found that altering the balance between histone methylation and demethylation impacted growth and proliferation. Of all genes tested, KDM3B, a histone H3K9 demethylase, was found to have the most antiproliferative effect. These results were phenocopied with a KDM3B CRISPR/Cas9 knockout. When tested in several PCa cell lines, the decrease in proliferation was remarkably specific to androgen-independent cells. Genetic rescue experiments showed that only the enzymatically active KDM3B could recover the phenotype. Surprisingly, despite the decreased proliferation of androgen-independent cell no alterations in the cell cycle distribution were observed following KDM3B knockdown. Whole transcriptome analyses revealed changes in the gene expression profile following loss of KDM3B, including downregulation of metabolic enzymes such as ARG2 and RDH11. Metabolomic analysis of KDM3B knockout showed a decrease in several critical amino acids. Overall, our work reveals, for the first time, the specificity and the dependence of KDM3B in CRPC proliferation.
Collapse
Affiliation(s)
- Hilal Saraç
- School of Medicine, Koç University, Istanbul, 34450, Turkey
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK
- Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Tunç Morova
- School of Medicine, Koç University, Istanbul, 34450, Turkey
- Vancouver Prostate Centre, University of British Columbia, Vancouver, V6H 3Z6, Canada
| | - Elisabete Pires
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK
| | - James McCullagh
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK
| | - Anıl Kaplan
- School of Medicine, Koç University, Istanbul, 34450, Turkey
| | - Ahmet Cingöz
- School of Medicine, Koç University, Istanbul, 34450, Turkey
| | | | - Tamer Önder
- School of Medicine, Koç University, Istanbul, 34450, Turkey
| | - Akane Kawamura
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK
- Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Nathan A Lack
- School of Medicine, Koç University, Istanbul, 34450, Turkey.
- Vancouver Prostate Centre, University of British Columbia, Vancouver, V6H 3Z6, Canada.
| |
Collapse
|
25
|
Design and Construction of a Focused DNA-Encoded Library for Multivalent Chromatin Reader Proteins. Molecules 2020; 25:molecules25040979. [PMID: 32098353 PMCID: PMC7070942 DOI: 10.3390/molecules25040979] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 02/11/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022] Open
Abstract
Chromatin structure and function, and consequently cellular phenotype, is regulated in part by a network of chromatin-modifying enzymes that place post-translational modifications (PTMs) on histone tails. These marks serve as recruitment sites for other chromatin regulatory complexes that ‘read’ these PTMs. High-quality chemical probes that can block reader functions of proteins involved in chromatin regulation are important tools to improve our understanding of pathways involved in chromatin dynamics. Insight into the intricate system of chromatin PTMs and their context within the epigenome is also therapeutically important as misregulation of this complex system is implicated in numerous human diseases. Using computational methods, along with structure-based knowledge, we have designed and constructed a focused DNA-Encoded Library (DEL) containing approximately 60,000 compounds targeting bi-valent methyl-lysine (Kme) reader domains. Additionally, we have constructed DNA-barcoded control compounds to allow optimization of selection conditions using a model Kme reader domain. We anticipate that this target-class focused approach will serve as a new method for rapid discovery of inhibitors for multivalent chromatin reader domains.
Collapse
|
26
|
Capasso M, Lasorsa VA, Cimmino F, Avitabile M, Cantalupo S, Montella A, De Angelis B, Morini M, de Torres C, Castellano A, Locatelli F, Iolascon A. Transcription Factors Involved in Tumorigenesis Are Over-Represented in Mutated Active DNA-Binding Sites in Neuroblastoma. Cancer Res 2019; 80:382-393. [PMID: 31784426 DOI: 10.1158/0008-5472.can-19-2883] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/24/2019] [Accepted: 11/22/2019] [Indexed: 11/16/2022]
Abstract
The contribution of coding mutations to oncogenesis has been largely clarified, whereas little is known about somatic mutations in noncoding DNA and their role in driving tumors remains controversial. Here, we used an alternative approach to interpret the functional significance of noncoding somatic mutations in promoting tumorigenesis. Noncoding somatic mutations of 151 neuroblastomas were integrated with ENCODE data to locate somatic mutations in regulatory elements specifically active in neuroblastoma cells, nonspecifically active in neuroblastoma cells, and nonactive. Within these types of elements, transcription factors (TF) were identified whose binding sites were enriched or depleted in mutations. For these TFs, a gene expression signature was built to assess their implication in neuroblastoma. DNA- and RNA-sequencing data were integrated to assess the effects of those mutations on mRNA levels. The pathogenicity of mutations was significantly higher in transcription factor binding site (TFBS) of regulatory elements specifically active in neuroblastoma cells, as compared with the others. Within these elements, there were 18 over-represented TFs involved mainly in cell-cycle phase transitions and 15 under-represented TFs primarily regulating cell differentiation. A gene expression signature based on over-represented TFs correlated with poor survival and unfavorable prognostic markers. Moreover, recurrent mutations in TFBS of over-represented TFs such as EZH2 affected MCF2L and ADP-ribosylhydrolase like 1 expression, among the others. We propose a novel approach to study the involvement of regulatory variants in neuroblastoma that could be extended to other cancers and provide further evidence that alterations of gene expression may have relevant effects in neuroblastoma development. SIGNIFICANCE: These findings propose a novel approach to study regulatory variants in neuroblastoma and suggest that noncoding somatic mutations have relevant implications in neuroblastoma development.
Collapse
Affiliation(s)
- Mario Capasso
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Napoli, Italy. .,CEINGE Biotecnologie Avanzate, Napoli, Italy.,IRCCS SDN, Napoli, Italy
| | - Vito Alessandro Lasorsa
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Napoli, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Flora Cimmino
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Napoli, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Marianna Avitabile
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Napoli, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy
| | | | - Annalaura Montella
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Napoli, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Biagio De Angelis
- Department of Pediatric Haematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Roma, Italy
| | - Martina Morini
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Carmen de Torres
- Developmental Tumor Biology Laboratory, Department of Oncology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Aurora Castellano
- Department of Pediatric Haematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Roma, Italy
| | - Franco Locatelli
- Department of Pediatric Haematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Roma, Italy.,Department of Paediatrics, Sapienza University of Rome, Roma, Italy
| | - Achille Iolascon
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Napoli, Italy. .,CEINGE Biotecnologie Avanzate, Napoli, Italy
| |
Collapse
|
27
|
Pang X, Xie R, Zhang Z, Liu Q, Wu S, Cui Y. Identification of SPP1 as an Extracellular Matrix Signature for Metastatic Castration-Resistant Prostate Cancer. Front Oncol 2019; 9:924. [PMID: 31620371 PMCID: PMC6760472 DOI: 10.3389/fonc.2019.00924] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/04/2019] [Indexed: 12/24/2022] Open
Abstract
Resistance to androgen deprivation therapy (ADT) is the main challenge for advanced fatal prostate cancer (PCa), which can gradually develop into metastatic castration-resistant prostate cancer (mCRPC). However, the pathologic mechanisms of mCRPC are still far from clear. Given the high incidence and mortality related to mCRPC, understanding the causes and pathogenesis of this condition as well as identifying potential biomarkers are of great importance. In the research reported here, we integrated several gene expression profiles from hormone sensitive prostate cancer (HSPC) and mCRPC datasets to identify differentially expressed genes (DEGs), key biological pathways, and cellular components. We found that extracellular matrix (ECM) genes were significantly enriched, and further filtered them using Pearson correlation analysis and stepwise regression to find ECM signatures to differentiate between the HSPC and mCRPC phenotypes. Six ECM signatures were input into K-nearest neighbor, logistic regression, naive Bayes, and random forest classifiers models. Random forest algorithm with the six-gene prognostic signatures showed best performance, which had high sensitivity and specificity for HSPC and mCRPC classification and further the six ECM signatures were validated in organoid models. Among the six ECM genes, SPP1 was identified as the key hub signature for PCa metastasis and drug resistance development; we found that both protein and mRNA expression levels of SPP1 were remarkably up-regulated in mCRPC compared with HSPC in organoid models and could regulate the androgen receptor signaling pathway. Therefore, SPP1 is a potential novel biomarker and therapeutic target for mCRPC. Further understanding of the role of SPP1 in mCRPC development may help to explore effectively therapeutic approaches for the prevention and intervention of drug resistance and metastasis.
Collapse
Affiliation(s)
- Xiaocong Pang
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| | - Ran Xie
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| | - Zhuo Zhang
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| | - Qianxin Liu
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| | - Shiliang Wu
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Yimin Cui
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| |
Collapse
|
28
|
Pang J, Wang L, Xu J, Xie Q, Liu Q, Tong D, Liu G, Huang Y, Yang X, Pan J, Yan X, Ma Q, Zhang D, Jiang J. A Renal Cell Carcinoma with Biallelic Somatic TSC2 Mutation: Clinical Study and Literature Review. Urology 2019; 133:96-102. [PMID: 31454656 DOI: 10.1016/j.urology.2019.08.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/07/2019] [Accepted: 08/10/2019] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To elucidate the effect of the biallelic somatic TSC2 mutations, identified in one adolescent patient, in renal cell carcinoma (RCC). METHODS Mutation analyses, immunohistochemistry and real-time polymerase chain reaction (PCR) were conducted. RESULTS Two novel somatic mutations of TSC2 in unilateral and solitary RCC samples from a 14-year-old female were identified. The pathological features suggest the tumor as a clear-cell renal cell carcinoma. In addition, immunohistochemistry revealed elevated levels of phosphorylated S6K1. Results from in vitro cellular experiments suggest that the mutant TSC2 proteins were quickly degraded and they failed to repress the phosphorylation of S6K1 and STAT3, which leads to constitutive activation of mTORC1 pathway and ultimately cause the development of RCC. CONCLUSION Detecting TSC2 mutation in patients with early RCC onset would be beneficial and mTOR inhibitor could be a therapeutic option for TSC2 mutation-induced RCC.
Collapse
Affiliation(s)
- Jian Pang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, PR China
| | - Linang Wang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, PR China
| | - Jing Xu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, PR China
| | - Qiubo Xie
- Department of Urology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, PR China
| | - Qiuli Liu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, PR China
| | - Dali Tong
- Department of Urology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, PR China
| | - Gaolei Liu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, PR China
| | - Yiqiang Huang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, PR China
| | - Xingxia Yang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, PR China
| | - Jinhong Pan
- Department of Urology, Southwest Hospital, Army Medical University, Chongqing, PR China
| | - Xiaochu Yan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, PR China
| | - Qiang Ma
- Department of Pathology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, PR China
| | - Dianzheng Zhang
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA
| | - Jun Jiang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, PR China.
| |
Collapse
|
29
|
McCann TS, Sobral LM, Self C, Hsieh J, Sechler M, Jedlicka P. Biology and targeting of the Jumonji-domain histone demethylase family in childhood neoplasia: a preclinical overview. Expert Opin Ther Targets 2019; 23:267-280. [PMID: 30759030 DOI: 10.1080/14728222.2019.1580692] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
INTRODUCTION Epigenetic mechanisms of gene regulatory control play fundamental roles in developmental morphogenesis, and, as more recently appreciated, are heavily implicated in the onset and progression of neoplastic disease, including cancer. Many epigenetic mechanisms are therapeutically targetable, providing additional incentive for understanding of their contribution to cancer and other types of neoplasia. Areas covered: The Jumonji-domain histone demethylase (JHDM) family exemplifies many of the above traits. This review summarizes the current state of knowledge of the functions and pharmacologic targeting of JHDMs in cancer and other neoplastic processes, with an emphasis on diseases affecting the pediatric population. Expert opinion: To date, the JHDM family has largely been studied in the context of normal development and adult cancers. In contrast, comparatively few studies have addressed JHDM biology in cancer and other neoplastic diseases of childhood, especially solid (non-hematopoietic) neoplasms. Encouragingly, the few available examples support important roles for JHDMs in pediatric neoplasia, as well as potential roles for JHDM pharmacologic inhibition in disease management. Further investigations of JHDMs in cancer and other types of neoplasia of childhood can be expected to both enlighten disease biology and inform new approaches to improve disease outcomes.
Collapse
Affiliation(s)
- Tyler S McCann
- a Department of Pathology , University of Colorado Denver, Anschutz Medical Campus , Aurora , CO , USA
| | - Lays M Sobral
- a Department of Pathology , University of Colorado Denver, Anschutz Medical Campus , Aurora , CO , USA
| | - Chelsea Self
- b Department of Pediatrics , University of Colorado Denver, Anschutz Medical Campus , Aurora , CO , USA
| | - Joseph Hsieh
- c Medical Scientist Training Program , University of Colorado Denver, Anschutz Medical Campus , Aurora , CO , USA
| | - Marybeth Sechler
- a Department of Pathology , University of Colorado Denver, Anschutz Medical Campus , Aurora , CO , USA.,d Cancer Biology Program , University of Colorado Denver, Anschutz Medical Campus , Aurora , CO , USA
| | - Paul Jedlicka
- a Department of Pathology , University of Colorado Denver, Anschutz Medical Campus , Aurora , CO , USA.,c Medical Scientist Training Program , University of Colorado Denver, Anschutz Medical Campus , Aurora , CO , USA.,d Cancer Biology Program , University of Colorado Denver, Anschutz Medical Campus , Aurora , CO , USA
| |
Collapse
|
30
|
Luo W, Wang Y. Hypoxia Mediates Tumor Malignancy and Therapy Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1136:1-18. [PMID: 31201713 DOI: 10.1007/978-3-030-12734-3_1] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hypoxia is a hallmark of the tumor microenvironment and contributes to tumor malignant phenotypes. Hypoxia-inducible factor (HIF) is a master regulator of intratumoral hypoxia and controls hypoxia-mediated pathological processes in tumors, including angiogenesis, metabolic reprogramming, epigenetic reprogramming, immune evasion, pH homeostasis, cell migration/invasion, stem cell pluripotency, and therapy resistance. In this book chapter, we reviewed the causes and types of intratumoral hypoxia, hypoxia detection methods, and the oncogenic role of HIF in tumorigenesis and chemo- and radio-therapy resistance.
Collapse
Affiliation(s)
- Weibo Luo
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA. .,Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Yingfei Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA. .,Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
31
|
Ying Z, Xiang G, Zheng L, Tang H, Duan L, Lin X, Zhao Q, Chen K, Wu Y, Xing G, Lv Y, Li L, Yang L, Bao F, Long Q, Zhou Y, He X, Wang Y, Gao M, Pei D, Chan WY, Liu X. Short-Term Mitochondrial Permeability Transition Pore Opening Modulates Histone Lysine Methylation at the Early Phase of Somatic Cell Reprogramming. Cell Metab 2018; 28:935-945.e5. [PMID: 30174306 DOI: 10.1016/j.cmet.2018.08.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 01/11/2018] [Accepted: 08/01/2018] [Indexed: 12/25/2022]
Abstract
Reprogramming of somatic cells to induced pluripotent stem cells reconfigures chromatin modifications. Whether and how this process is regulated by signals originating in the mitochondria remain unknown. Here we show that the mitochondrial permeability transition pore (mPTP), a key regulator of mitochondrial homeostasis, undergoes short-term opening during the early phase of reprogramming and that this transient activation enhances reprogramming. In mouse embryonic fibroblasts, greater mPTP opening correlates with higher reprogramming efficiency. The reprogramming-promoting function of mPTP opening is mediated by plant homeodomain finger protein 8 (PHF8) demethylation of H3K9me2 and H3K27me3, leading to reduction in their occupancies at the promoter regions of pluripotency genes. mPTP opening increases PHF8 protein levels downstream of mitochondrial reactive oxygen species production and miR-101c and simultaneously elevates levels of PHF8's cofactor, α-ketoglutarate. Our findings represent a novel mitochondria-to-nucleus pathway in cell fate determination by mPTP-mediated epigenetic regulation.
Collapse
Affiliation(s)
- Zhongfu Ying
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Ge Xiang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Lingjun Zheng
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China; Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| | - Haite Tang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Lifan Duan
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xiaobing Lin
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Qiuge Zhao
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Keshi Chen
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yi Wu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Guangsuo Xing
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China; Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| | - Yiwang Lv
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Linpeng Li
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Liang Yang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Feixiang Bao
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Qi Long
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yanshuang Zhou
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xueying He
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yaofeng Wang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Minghui Gao
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Duanqing Pei
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Wai-Yee Chan
- CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, China
| | - Xingguo Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou 510530, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Guangzhou 510530, China.
| |
Collapse
|
32
|
Yu Y, Zhang Q, Ma C, Yang X, Lin R, Zhang H, Liu Y, Han Z, Cheng J. Mesenchymal stem cells recruited by castration-induced inflammation activation accelerate prostate cancer hormone resistance via chemokine ligand 5 secretion. Stem Cell Res Ther 2018; 9:242. [PMID: 30257726 PMCID: PMC6158918 DOI: 10.1186/s13287-018-0989-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 07/31/2018] [Accepted: 08/20/2018] [Indexed: 12/20/2022] Open
Abstract
Background Androgen deprivation (AD) as the first-line treatment for advanced prostate cancer (PCa) is insufficient for a long-term effect. Castration resistance remains the greatest obstacle in PCa clinical therapy. Mesenchymal stem cells (MSCs) can migrate into PCa tissues contributing to tumor progression, therefore, in this study we explored the effect of AD on MSC migration to PCa and elicited its importance for the emergence of castration resistance. Methods MSC migration assay was performed in several PCa cells (LNCaP, VCaP, and 22Rv1) using in-vivo and in-vitro approaches. Reactive oxygen species generation was evaluated by fluorescence assay. IL-1β was analyzed by immunohistochemistry, and neutralization experiments were conducted using neutralization antibody. Stem markers (CD133, CD44, and SOX2) were quantified by real-time PCR analysis. The concentration of chemokine ligand 5 was measured by enzyme-linked immunosorbent assay and small hairpin RNA was used for functional analyses. Results AD could significantly contribute to PCa recruitment of MSCs in vivo and in vitro. AD-induced oxidative stress could promote the inflammatory response mediated by IL-1β secretion via activating the NF-κB signaling pathway. Moreover, N-acetylcysteine could significantly inhibit MSC recruitment to PCa sites when AD is performed. Furthermore, we found MSCs could increase stemness of PCa cells via promoting chemokine ligand 5 secretion in the AD condition, and consequently accelerate emergence of castration resistance. Conclusions Our results suggest that castration in clinical PCa therapy may elicit oxidative stress in tumor sites, resulting in increased MSC migration and in tumor cell growth in an androgen-independent manner. Blocking MSC migration to the tumor may provide a new potential target to suppress castration-resistant PCa emergence. Electronic supplementary material The online version of this article (10.1186/s13287-018-0989-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yang Yu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Qingyun Zhang
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Chengzhong Ma
- Department of Urology, Guangxi International Zhuang Medicine Hospital, Nanning, 530021, China
| | - Xue Yang
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai, 200438, China
| | - Rui Lin
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Hongxiang Zhang
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Yan Liu
- The Fifth Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Zhipeng Han
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai, 200438, China
| | - Jiwen Cheng
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, China.
| |
Collapse
|
33
|
Zhou W, Gong L, Wu Q, Xing C, Wei B, Chen T, Zhou Y, Yin S, Jiang B, Xie H, Zhou L, Zheng S. PHF8 upregulation contributes to autophagic degradation of E-cadherin, epithelial-mesenchymal transition and metastasis in hepatocellular carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:215. [PMID: 30180906 PMCID: PMC6122561 DOI: 10.1186/s13046-018-0890-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 08/21/2018] [Indexed: 12/12/2022]
Abstract
Background Plant homeodomain finger protein 8 (PHF8) serves an activator of epithelial-mesenchymal transition (EMT) and is implicated in various tumors. However, little is known about PHF8 roles in hepatocellular carcinoma (HCC) and regulating E-cadherin expression. Methods PHF8 expression pattern was investigated by informatic analysis and verified by RT-qPCR and immunochemistry in HCC tissues and cell lines. CCK8, xenograft tumor model, transwell assay, and tandem mCherry-GFP-LC3 fusion protein assay were utilized to assess the effects of PHF8 on proliferation, metastasis and autophagy of HCC cells in vitro and in vivo. ChIP, immunoblot analysis, rescue experiments and inhibitor treatment were used to clarify the mechanism by which PHF8 facilitated EMT, metastasis and autophagy. Results PHF8 upregulation was quite prevalent in HCC tissues and closely correlated with worse overall survival and disease-relapse free survival. Furthermore, PHF8-knockdown dramatically suppressed cell growth, migration, invasion and autophagy, and the expression of SNAI1, VIM, N-cadherin and FIP200, and increased E-cadherin level, while PHF8-overexpression led to the opposite results. Additionally, FIP200 augmentation reversed the inhibited effects of PHF8-siliencing on tumor migration, invasion and autophagy. Mechanistically, PHF8 was involved in transcriptionally regulating the expression of SNAI1, VIM and FIP200, rather than N-cadherin and E-cadherin. Noticeably, E-cadherin degradation could be accelerated by PHF8-mediated FIP200-dependent autophagy, a crucial pathway complementary to transcriptional repression of E-cadherin by SNAI1 activation. Conclusion These findings suggested that PHF8 played an oncogenic role in facilitating FIP200-dependent autophagic degradation of E-cadherin, EMT and metastasis in HCC. PHF8 might be a promising target for prevention, treatment and prognostic prediction of HCC. Electronic supplementary material The online version of this article (10.1186/s13046-018-0890-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wuhua Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ transplantation, CAMS, Hangzhou, China.,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang Province, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Disease, Zhejiang University, Hangzhou, China
| | - Li Gong
- Department of Endocrinology, Taihe Hospital, Shiyan, China
| | - Qinchuan Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ transplantation, CAMS, Hangzhou, China.,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang Province, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Disease, Zhejiang University, Hangzhou, China
| | - Chunyang Xing
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bajin Wei
- NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ transplantation, CAMS, Hangzhou, China.,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang Province, China
| | - Tianchi Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ transplantation, CAMS, Hangzhou, China.,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang Province, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Disease, Zhejiang University, Hangzhou, China
| | - Yuan Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ transplantation, CAMS, Hangzhou, China.,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang Province, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Disease, Zhejiang University, Hangzhou, China
| | - Shengyong Yin
- NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ transplantation, CAMS, Hangzhou, China.,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang Province, China
| | - Bin Jiang
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Shiyan, China
| | - Haiyang Xie
- NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China.,Key Laboratory of the Diagnosis and Treatment of Organ transplantation, CAMS, Hangzhou, China.,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang Province, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Disease, Zhejiang University, Hangzhou, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China. .,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China. .,Key Laboratory of the Diagnosis and Treatment of Organ transplantation, CAMS, Hangzhou, China. .,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang Province, China. .,Collaborative Innovation Center for Diagnosis Treatment of Infectious Disease, Zhejiang University, Hangzhou, China.
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China. .,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China. .,Key Laboratory of the Diagnosis and Treatment of Organ transplantation, CAMS, Hangzhou, China. .,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang Province, China. .,Collaborative Innovation Center for Diagnosis Treatment of Infectious Disease, Zhejiang University, Hangzhou, China.
| |
Collapse
|
34
|
Liu Q, Tong D, Liu G, Gao J, Wang LA, Xu J, Yang X, Xie Q, Huang Y, Pang J, Wang L, He Y, Zhang D, Ma Q, Lan W, Jiang J. Metformin Inhibits Prostate Cancer Progression by Targeting Tumor-Associated Inflammatory Infiltration. Clin Cancer Res 2018; 24:5622-5634. [PMID: 30012567 DOI: 10.1158/1078-0432.ccr-18-0420] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/21/2018] [Accepted: 07/10/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Inflammatory infiltration plays important roles in both carcinogenesis and metastasis. We are interested in understanding the inhibitory mechanism of metformin on tumor-associated inflammation in prostate cancer.Experimental Design: By using a transgenic adenocarcinoma of the mouse prostate (TRAMP) mouse model, in vitro macrophage migration assays, and patient samples, we examined the effect of metformin on tumor-associated inflammation during the initiation and after androgen deprivation therapy of prostate cancer.Results: Treating TRAMP mice with metformin delays prostate cancer progression from low-grade prostatic intraepithelial neoplasia to high-grade PIN, undifferentiated to well-differentiated, and PIN to adenocarcinoma with concurrent inhibition of inflammatory infiltration evidenced by reduced recruitment of macrophages. Furthermore, metformin is capable of inhibiting the following processes: inflammatory infiltration after androgen deprivation therapy (ADT) induced by surgically castration in mice, bicalutamide treatment in patients, and hormone deprivation in LNCaP cells. Mechanistically, metformin represses inflammatory infiltration by downregulating both COX2 and PGE2 in tumor cells.Conclusions: Metformin is capable of repressing prostate cancer progression by inhibiting infiltration of tumor-associated macrophages, especially those induced by ADT, by inhibiting the COX2/PGE2 axis, suggesting that a combination of ADT with metformin could be a more efficient therapeutic strategy for prostate cancer treatment. Clin Cancer Res; 24(22); 5622-34. ©2018 AACR.
Collapse
Affiliation(s)
- Qiuli Liu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Dali Tong
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Gaolei Liu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Jie Gao
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Lin-Ang Wang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Jing Xu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Xingxia Yang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Qiubo Xie
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Yiqiang Huang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Jian Pang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Luofu Wang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Yong He
- Department of Respiratory Medicine, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Dianzheng Zhang
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania
| | - Qiang Ma
- Department of Pathology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Weihua Lan
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China.
| | - Jun Jiang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China.
| |
Collapse
|
35
|
Licon-Munoz Y, Fordyce CA, Hayek SR, Parra KJ. V-ATPase-dependent repression of androgen receptor in prostate cancer cells. Oncotarget 2018; 9:28921-28934. [PMID: 29988966 PMCID: PMC6034745 DOI: 10.18632/oncotarget.25641] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/04/2018] [Indexed: 12/17/2022] Open
Abstract
Prostate Cancer (PCa) is the most commonly diagnosed cancer and the third leading cause of death for men in the United States. Suppression of androgen receptor (AR) expression is a desirable mechanism to manage PCa. Our studies showed that AR expression was reduced in LAPC4 and LNCaP PCa cell lines treated with nanomolar concentrations of the V-ATPase inhibitor concanamycin A (CCA). This treatment decreased PSA mRNA levels, indicative of reduced AR activity. V-ATPase-dependent repression of AR expression was linked to defective endo-lysosomal pH regulation and reduced AR expression at the transcriptional level. CCA treatment increased the protein level and nuclear localization of the alpha subunit of the transcription factor HIF-1 (HIF-1α) in PCa cells via decreased hydroxylation and degradation of HIF-1α. The addition of iron (III) citrate restored HIF-1α hydroxylation and decreased total HIF-1α levels in PCa cells treated with CCA. Moreover, iron treatment partially rescued CCA-mediated AR repression. Dimethyloxalylglycine (DMOG), which prevents HIF-1α degradation independently of V-ATPase, also decreased AR levels, supporting our hypothesis that HIF-1α serves as a downstream mediator in the V-ATPase-AR axis. We propose a new V-ATPase-dependent mechanism to inhibit androgen receptor expression in prostate cancer cells involving defective endosomal trafficking of iron and the inhibition of HIF-1 α-subunit turnover.
Collapse
Affiliation(s)
- Yamhilette Licon-Munoz
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Colleen A Fordyce
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Summer Raines Hayek
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Karlett J Parra
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| |
Collapse
|
36
|
Zhou H, Wu G, Ma X, Xiao J, Yu G, Yang C, Xu N, Zhang B, Zhou J, Ye Z, Wang Z. Attenuation of TGFBR2 expression and tumour progression in prostate cancer involve diverse hypoxia-regulated pathways. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:89. [PMID: 29699590 PMCID: PMC5921809 DOI: 10.1186/s13046-018-0764-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 04/16/2018] [Indexed: 12/19/2022]
Abstract
Background Dysregulation of transforming growth factor β (TGF-β) signaling and hypoxic microenvironment have respectively been reported to be involved in disease progression in malignancies of prostate. Emerging evidence indicates that downregulation of TGFBR2, a pivotal regulator of TGF-β signaling, may contribute to carcinogenesis and progression of prostate cancer (PCa). However, the biological function and regulatory mechanism of TGFBR2 in PCa remain poorly understood. In this study, we propose to investigate the crosstalk of hypoxia and TGF-β signaling and provide insight into the molecular mechanism underlying the regulatory pathways in PCa. Methods Prostate cancer cell lines were cultured in hypoxia or normoxia to evaluate the effect of hypoxia on TGFBR2 expression. Methylation specific polymerase chain reaction (MSP) and demethylation agents was used to evaluate the methylation regulation of TGFBR2 promoter. Besides, silencing of EZH2 via specific siRNAs or chemical inhibitor was used to validate the regulatory effect of EZH2 on TGFBR2. Moreover, we conducted PCR, western blot, and luciferase assays which studied the relationship of miR-93 and TGFBR2 in PCa cell lines and specimens. We also detected the impacts of hypoxia on EZH2 and miR-93, and further examined the tumorigenic functions of miR-93 on proliferation and epithelial-mesenchymal transition via a series of experiments. Results TGFBR2 expression was attenuated under hypoxia. Hypoxia-induced EZH2 promoted H3K27me3 which caused TGFBR2 promoter hypermethylation and contributed to its epigenetic silencing in PCa. Besides, miR-93 was significantly upregulated in PCa tissues and cell lines, and negatively correlated with the expression of TGFBR2. Ectopic expression of miR-93 promoted cell proliferation, migration and invasion in PCa, and its expression could also be induced by hypoxia. In addition, TGFBR2 was identified as a bona fide target of miR-93. Conclusions Our findings elucidate diverse hypoxia-regulated pathways including EZH2-mediated hypermethylation and miR-93-induced silencing contribute to attenuation of TGFBR2 expression and promote cancer progression in prostate cancer. Electronic supplementary material The online version of this article (10.1186/s13046-018-0764-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hui Zhou
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guanqing Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Urology, Aerospace Center Hospital(ASCH), Beijing, 100076, China
| | - Xueyou Ma
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun Xiao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gan Yu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chunguang Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Nan Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bao Zhang
- Department of Urology, Aerospace Center Hospital(ASCH), Beijing, 100076, China
| | - Jun Zhou
- Department of Urology, The third people Hospital of Hubei Province, Wuhan, 430030, China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhihua Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
37
|
Ruggero K, Farran-Matas S, Martinez-Tebar A, Aytes A. Epigenetic Regulation in Prostate Cancer Progression. ACTA ACUST UNITED AC 2018; 4:101-115. [PMID: 29888169 PMCID: PMC5976687 DOI: 10.1007/s40610-018-0095-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Purpose of Review An important number of newly identified molecular alterations in prostate cancer affect gene encoding master regulators of chromatin biology epigenetic regulation. This review will provide an updated view of the key epigenetic mechanisms underlying prostate cancer progression, therapy resistance, and potential actionable mechanisms and biomarkers. Recent Findings Key players in chromatin biology and epigenetic master regulators has been recently described to be crucially altered in metastatic CRPC and tumors that progress to AR independency. As such, epigenetic dysregulation represents a driving mechanism in the reprograming of prostate cancer cells as they lose AR-imposed identity. Summary Chromatin integrity and accessibility for transcriptional regulation are key features altered in cancer progression, and particularly relevant in nuclear hormone receptor-driven tumors like prostate cancer. Understanding how chromatin remodeling dictates prostate development and how its deregulation contributes to prostate cancer onset and progression may improve risk stratification and treatment selection for prostate cancer patients.
Collapse
Affiliation(s)
- Katia Ruggero
- Programs of Molecular Mechanisms and Experimental Therapeutics in Oncology (ONCOBell), Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research, Granvia de l'Hopitalet, 199 08908, L'Hospitalet de Llobregat, 08907 Barcelona, Spain
| | - Sonia Farran-Matas
- Programs of Molecular Mechanisms and Experimental Therapeutics in Oncology (ONCOBell), Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research, Granvia de l'Hopitalet, 199 08908, L'Hospitalet de Llobregat, 08907 Barcelona, Spain
| | - Adrian Martinez-Tebar
- Programs of Molecular Mechanisms and Experimental Therapeutics in Oncology (ONCOBell), Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research, Granvia de l'Hopitalet, 199 08908, L'Hospitalet de Llobregat, 08907 Barcelona, Spain
| | - Alvaro Aytes
- Programs of Molecular Mechanisms and Experimental Therapeutics in Oncology (ONCOBell), Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research, Granvia de l'Hopitalet, 199 08908, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.,Programs of Cancer Therapeutics Resistance (ProCURE), Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, 08907 Barcelona, Spain
| |
Collapse
|
38
|
Daskalaki MG, Tsatsanis C, Kampranis SC. Histone methylation and acetylation in macrophages as a mechanism for regulation of inflammatory responses. J Cell Physiol 2018; 233:6495-6507. [PMID: 29574768 DOI: 10.1002/jcp.26497] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/22/2018] [Indexed: 12/25/2022]
Abstract
Macrophages respond to noxious stimuli and contribute to inflammatory responses by eliminating pathogens or damaged tissue and maintaining homeostasis. Response to activation signals and maintenance of homeostasis require tight regulation of genes involved in macrophage activation and inactivation processes, as well as genes involved in determining their polarization state. Recent evidence has revealed that such regulation occurs through histone modifications that render inflammatory or polarizing gene promoters accessible to transcriptional complexes. Thus, inflammatory and anti-inflammatory genes are regulated by histone acetylation and methylation, determining their activation state. Herein, we review the current knowledge on the role of histone modifying enzymes (acetyltransferases, deacetylases, methyltransferases, and demethylases) in determining the responsiveness and M1 or M2 polarization of macrophages. The contribution of these enzymes in the development of inflammatory diseases is also presented.
Collapse
Affiliation(s)
- Maria G Daskalaki
- Laboratory of Biochemistry, Medical School, University of Crete, Heraklion, Crete, Greece.,Laboratory of Clinical Chemistry, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Christos Tsatsanis
- Laboratory of Clinical Chemistry, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Sotirios C Kampranis
- Laboratory of Biochemistry, Medical School, University of Crete, Heraklion, Crete, Greece
| |
Collapse
|
39
|
The histone demethylase PHF8 promotes adult acute lymphoblastic leukemia through interaction with the MEK/ERK signaling pathway. Biochem Biophys Res Commun 2018; 496:981-987. [PMID: 29330049 DOI: 10.1016/j.bbrc.2018.01.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 01/08/2018] [Indexed: 01/21/2023]
Abstract
Adult acute lymphoblastic leukemia (ALL) is a malignant disorder of lymphoid progenitor cells that is associated with a high risk of relapse and poor prognosis. Thus, novel pathogenic mechanisms and therapeutic targets need to be explored. Histone methylation is one of the most significant chromatin post-translational modifications. Here, we show that the histone demethylase PHF8 is highly expressed in a large number of ALL clinical specimens and that PHF8 expression is associated with ALL progression. PHF8 knockdown inhibits proliferation and promotes the apoptosis of ALL cells in vitro as well as attenuates tumor growth in vivo. PHF8 transcriptionally upregulates MEK1, a key molecule in the MEK/ERK pathway, at least partially by directly binding to its promoter, thereby activating the MEK/ERK pathway. In addition, we found that an inhibitor of the MEK/ERK pathway, PD184352, subsequently suppresses PHF8 expression. Thus, PHF8 forms a positive feedback loop with the MEK/ERK pathway, and PHF8 knockdown enhances the lethality of PD184352 in ALL cells. In conclusion, this study identifies oncogenic functions of PHF8 in adult ALL and suggests a novel epigenetic strategy for disease intervention.
Collapse
|
40
|
Vlachostergios PJ, Paddock M, Molina AM. Molecular Targeted Therapies of Prostate Cancer. MOLECULAR PATHOLOGY LIBRARY 2018. [DOI: 10.1007/978-3-319-64096-9_29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
41
|
Liu Q, Tong D, Liu G, Xu J, Do K, Geary K, Zhang D, Zhang J, Zhang Y, Li Y, Bi G, Lan W, Jiang J. Metformin reverses prostate cancer resistance to enzalutamide by targeting TGF-β1/STAT3 axis-regulated EMT. Cell Death Dis 2017; 8:e3007. [PMID: 28837141 PMCID: PMC5596596 DOI: 10.1038/cddis.2017.417] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 07/14/2017] [Accepted: 07/24/2017] [Indexed: 02/07/2023]
Abstract
Although the newly developed second-generation anti-androgen drug enzalutamide can repress prostate cancer progression significantly, it only extends the survival of prostate cancer patients by 4–6 months mainly due to the occurrence of enzalutamide resistance. Most of the previous studies on AR antagonist resistance have been focused on AR signaling. Therefore, the non-AR pathways on enzalutamide resistance remain largely unknown. By using C4-2, CWR22Rv1 and LNCaP cell lines, as well as mice bearing CWR22Rv1 xenografts treated with either enzalutamide or metformin alone or in combination, we demonstrated that metformin is capable of reversing enzalutamide resistance and restores sensitivity of CWR22Rv1 xenografts to enzalutamide. We showed that metformin alleviated resistance to enzalutamide by inhibiting EMT. Furthermore, based on the effect of metformin on the activation of STAT3 and expression of TGF-β1, we propose that metformin exerts its effects by targeting the TGF-β1/STAT3 axis. These findings suggest that combination of metformin with enzalutamide could be a more efficacious therapeutic strategy for the treatment of castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Qiuli Liu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Dali Tong
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Gaolei Liu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Jing Xu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Khang Do
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA 19131, USA
| | - Kyla Geary
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA 19131, USA
| | - Dianzheng Zhang
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA 19131, USA
| | - Jun Zhang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Yao Zhang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Yaoming Li
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Gang Bi
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Weihua Lan
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Jun Jiang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| |
Collapse
|
42
|
Maina PK, Shao P, Jia X, Liu Q, Umesalma S, Marin M, Long D, Concepción-Román S, Qi HH. Histone demethylase PHF8 regulates hypoxia signaling through HIF1α and H3K4me3. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:1002-1012. [PMID: 28734980 PMCID: PMC5776039 DOI: 10.1016/j.bbagrm.2017.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 07/10/2017] [Accepted: 07/18/2017] [Indexed: 11/12/2022]
Abstract
Hypoxia through transcription factor HIF1α plays a critical role in cancer development. In prostate cancer, HIF1α interplays with androgen receptor (AR) to contribute to the progression of this disease to its lethal form—castration-resistant prostate cancer (CRPC). Hypoxia upregulates several epigenetic factors including histone demethylase KDM3A which is a critical co-factor of HIF1α. However, how histone demethylases regulate hypoxia signaling is not fully understood. Here, we report that histone demethylase PHF8 plays an essential role in hypoxia signaling. Knockdown or knockout of PHF8 by RNAi or CRISPR-Cas9 system reduced the activation of HIF1α and the induction of HIF1α target genes including KDM3A. Mechanistically, PHF8 regulates hypoxia inducible genes mainly through sustaining the level of trimethylated histone 3 lysine 4 (H3K4me3), an active mark in transcriptional regulation. The positive role of PHF8 in hypoxia signaling extended to hypoxia-induced neuroendocrine differentiation (NED), wherein PHF8 cooperates with KDM3A to regulate the expression of NED genes. Moreover, we discovered that the role of PHF8 in hypoxia signaling is associated with the presence of full-length AR in CRPC cells. Collectively, our study identified PHF8 as a novel epigenetic factor in hypoxia signaling, and the underlying regulatory mechanisms likely apply to general cancer development involving HIF1α. Therefore, targeting PHF8 can potentially be a novel therapeutic strategy in cancer therapy.
Collapse
Affiliation(s)
- Peterson Kariuki Maina
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Peng Shao
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Xiongfei Jia
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Qi Liu
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Shaikamjad Umesalma
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Maximo Marin
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Donald Long
- Department of Biology, Southern Utah University, Cedar City, UT 84720, USA
| | | | - Hank Heng Qi
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA.
| |
Collapse
|
43
|
Rasool RU, Nayak D, Chakraborty S, Faheem MM, Rah B, Mahajan P, Gopinath V, Katoch A, Iqra Z, Yousuf SK, Mukherjee D, Kumar LD, Nargotra A, Goswami A. AKT is indispensable for coordinating Par-4/JNK cross talk in p21 downmodulation during ER stress. Oncogenesis 2017; 6:e341. [PMID: 28530706 PMCID: PMC5523074 DOI: 10.1038/oncsis.2017.41] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/22/2017] [Accepted: 04/17/2017] [Indexed: 12/25/2022] Open
Abstract
The double-edged role of p21 to command survival and apoptosis is emerging. The current investigation highlights ER stress-mediated JNK activation that plausibly triggers cell death by attenuating endogenous p21 level. Here, we demonstrated that ER stress activator 3-AWA diminishes the p21 levels in cancer cells by averting the senescent phenotype to commence G2/M arrest. In essence, the deceleration in p21 level occurs through ER stress/JNK/Caspase-3 axis via activation/induction of proapoptotic Par-4 and inhibition of AKT. The molecular dynamics studies identified important interactions, which may be responsible for the AKT inhibition and efficacy of 3-AWA towards AKT binding pocket. Interestingly, the p21 deceleration was rescued by incubating the cells with 3-AWA in the presence of an ER stress inhibitor, Salubrinal. Furthermore, we demonstrated that p21 expression decreases solitarily in Par-4+/+ MEFs; albeit, ER stress-induced JNK activation was observed in both Par-4+/+ and Par-4−/− MEFs. Par-4 knockdown or overexpression studies established that ectopic Par-4 along with ER stress are not sufficient to downregulate p21 in PC-3 cells but are adequate for DU-145 cells and that the ER stress inflicted activation of JNK, inhibition of AKT and Par-4 induction are all crucial to p21 downmodulation by 3-AWA. By using isogenic cell lines, such as HCT-116 p53+/+ and HCT-116 p53−/−, we found that deceleration in p21 expression due to ER stress is p53 independent. Moreover, in orthotopic carcinogen-induced rat colorectal carcinoma model, we found that 3-AWA inhibits colorectal tumor growth and formation of colorectal polyps at a tolerable dose, similar to the first-line drug for colorectal cancer-5-fluorouracil.
Collapse
Affiliation(s)
- R U Rasool
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - D Nayak
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - S Chakraborty
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - M M Faheem
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - B Rah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
| | - P Mahajan
- Discovery Informatics Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - V Gopinath
- Cancer Biology Division, CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - A Katoch
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Z Iqra
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - S K Yousuf
- Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - D Mukherjee
- Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - L D Kumar
- Cancer Biology Division, CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - A Nargotra
- Discovery Informatics Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - A Goswami
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| |
Collapse
|
44
|
Baumgart SJ, Haendler B. Exploiting Epigenetic Alterations in Prostate Cancer. Int J Mol Sci 2017; 18:ijms18051017. [PMID: 28486411 PMCID: PMC5454930 DOI: 10.3390/ijms18051017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/04/2017] [Accepted: 05/04/2017] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer affects an increasing number of men worldwide and is a leading cause of cancer-associated deaths. Beside genetic mutations, many epigenetic alterations including DNA and histone modifications have been identified in clinical prostate tumor samples. They have been linked to aberrant activity of enzymes and reader proteins involved in these epigenetic processes, leading to the search for dedicated inhibitory compounds. In the wake of encouraging anti-tumor efficacy results in preclinical models, epigenetic modulators addressing different targets are now being tested in prostate cancer patients. In addition, the assessment of microRNAs as stratification biomarkers, and early clinical trials evaluating suppressor microRNAs as potential prostate cancer treatment are being discussed.
Collapse
Affiliation(s)
- Simon J Baumgart
- Drug Discovery, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany.
| | - Bernard Haendler
- Drug Discovery, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany.
| |
Collapse
|
45
|
Mittal K, Donthamsetty S, Kaur R, Yang C, Gupta MV, Reid MD, Choi DH, Rida PCG, Aneja R. Multinucleated polyploidy drives resistance to Docetaxel chemotherapy in prostate cancer. Br J Cancer 2017; 116:1186-1194. [PMID: 28334734 PMCID: PMC5418452 DOI: 10.1038/bjc.2017.78] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 02/24/2017] [Accepted: 03/01/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Docetaxel is the only FDA-approved first-line treatment for castration-resistant prostate cancer (CRPC) patients. Docetaxel treatment inevitably leads to tumour recurrence after an initial therapeutic response with generation of multinucleated polyploid (MP) cells. Here we investigated role of MP cells in clinical relapse of CRPC. METHODS Prostate cancer (PC-3) cells were treated with docetaxel (5 nM) for 3 days followed by a washout and samples were collected at close intervals over 35 days post drug washout. The tumorigenic potential of the giant MP cells was studied by implanting MP cells subcutaneously as tumour xenografts in nude mice. RESULTS Docetaxel-induced polyploid cells undergo mitotic slippage and eventually spawn mononucleated cells via asymmetric cell division or neosis. Both MP and cells derived from polyploid cells had increased survival signals, were positive for CD44 and were resistant to docetaxel chemotherapy. Although MP cells were tumorigenic in nude mice, these cells took a significantly longer time to form tumours compared with parent PC-3 cells. CONCLUSIONS Generation of MP cells upon docetaxel therapy is an adaptive response of apoptosis-reluctant cells. These giant cells ultimately contribute to the generation of mononucleated aneuploid cells via neosis and may have a fundamental role precipitating clinical relapse and chemoresistance in CRPC.
Collapse
Affiliation(s)
- Karuna Mittal
- Department of Biology, Georgia State University, Atlanta, GA-30303, USA
| | | | - Ramneet Kaur
- Department of Biology, Georgia State University, Atlanta, GA-30303, USA
| | - Chunhua Yang
- Department of Biology, Georgia State University, Atlanta, GA-30303, USA
| | | | - Michelle D Reid
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Da Hoon Choi
- Department of Biology, Georgia State University, Atlanta, GA-30303, USA
| | - Padmashree C G Rida
- Department of Biology, Georgia State University, Atlanta, GA-30303, USA.,Novazoi Theranostics, Inc., Rolling Hills Estates, CA 90274, USA
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA-30303, USA
| |
Collapse
|
46
|
Li S, Sun A, Liang X, Ma L, Shen L, Li T, Zheng L, Shang W, Zhao W, Jia J. Histone demethylase PHF8 promotes progression and metastasis of gastric cancer. Am J Cancer Res 2017; 7:448-461. [PMID: 28401003 PMCID: PMC5385635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 02/15/2017] [Indexed: 06/07/2023] Open
Abstract
Histone demethylase plant homeodomain (PHD) finger protein 8 (PHF8) has been implicated in tumor development and malignant progression in various types of cancers. However, its potential roles in gastric cancer (GC) have not been explored. In this report, we show that PHF8 expression is upregulated in GC tissues, and the enhanced PHF8 level indicates a poor prognosis of GC patients. PHF8 knockdown reduces proliferation and metastasis of GC cells, while PHF8 overexpression has the opposite effects. Mechanistically, PHF8 interacts with β-catenin, and binds to the promoter region of vimentin, leading to the promotion of vimentin transcription. In addition, we show that H. pylori, the single most important risk factor for GC, markedly induce PHF8 expression. Our results suggest that H. pylori-induced PHF8-β-catenin-vimentin axis activation is a novel mechanism for GC malignant progression. Thus, we identify PHF8 as an oncogenic factor of GC, and suggest PHF8 might be a potential molecular target for therapeutic approaches for GC.
Collapse
Affiliation(s)
- Shuyan Li
- Department of Microbiology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong UniversityJinan 250012, Shandong, PR China
| | - Ao Sun
- Department of Microbiology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong UniversityJinan 250012, Shandong, PR China
| | - Xiuming Liang
- Department of Microbiology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong UniversityJinan 250012, Shandong, PR China
| | - Lin Ma
- Department of Microbiology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong UniversityJinan 250012, Shandong, PR China
| | - Li Shen
- Department of Microbiology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong UniversityJinan 250012, Shandong, PR China
| | - Tongyu Li
- Department of Microbiology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong UniversityJinan 250012, Shandong, PR China
| | - Lixin Zheng
- Department of Microbiology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong UniversityJinan 250012, Shandong, PR China
| | - Wenjing Shang
- Department of Microbiology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong UniversityJinan 250012, Shandong, PR China
| | - Wei Zhao
- Department of Immunology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong UniversityJinan 250012, Shandong, PR China
| | - Jihui Jia
- Department of Microbiology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong UniversityJinan 250012, Shandong, PR China
| |
Collapse
|